Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrew R. Zolopa is active.

Publication


Featured researches published by Andrew R. Zolopa.


AIDS | 2000

Adherence to protease inhibitors, Hiv-1 viral load, and development of drug resistance in an indigent population

David R. Bangsberg; Frederick Hecht; Edwin D. Charlebois; Andrew R. Zolopa; Mark Holodniy; Lewis B. Sheiner; Joshua D. Bamberger; Margaret A. Chesney; Andrew R. Moss

ObjectiveTo examine the relationship between adherence, viral suppression and antiretroviral resistance in HIV-infected homeless and marginally housed people on protease inhibitor (PI) therapy. Design and settingA cross-sectional analysis of subjects in an observational prospective cohort systematically sampled from free meal lines, homeless shelters and low-income, single-room occupancy (SRO) hotels. ParticipantsThirty-four HIV-infected people with a median of 12 months of PI therapy. Main outcomesAdherence measured by periodic unannounced pill counts, electronic medication monitoring, and self-report; HIV RNA viral load; and HIV-1 genotypic changes associated with drug resistance. ResultsMedian adherence was 89, 73, and 67% by self-report, pill count, and electronic medication monitor, respectively. Thirty-eight per cent of the population had over 90% adherence by pill count. Depending on the measure, adherence explained 36–65% of the variation in concurrent HIV RNA levels. The three adherence measures were closely related. Of 20 genotyped patients who received a new reverse transcriptase inhibitor (RTI) when starting a PI, three had primary protease gene substitutions. Of 12 genotyped patients who received a PI without a new RTI, six had primary protease gene substitutions (P < 0.03). ConclusionA substantial proportion of homeless and marginally housed individuals had good adherence to PI therapy. A strong relationship was found between independent methods of measuring adherence and concurrent viral suppression. PI resistance was more closely related to the failure to change RTI when starting a PI than to the level of adherence.


AIDS | 2001

Non-adherence to highly active antiretroviral therapy predicts progression to AIDS

David R. Bangsberg; Sharon Perry; Edwin D. Charlebois; Richard A.F. Clark; Marjorie Roberston; Andrew R. Zolopa; Andrew R. Moss

The introduction of highly active antiretroviral therapy (HAART) has produced a dramatic reduction in mortality among HIV-infected individuals [1–4]. Whereas the level of adherence to HAART is closely associated with suppression of the HIV viral load in plasma [5–14], a relationship between adherenc


PLOS ONE | 2009

Early Antiretroviral Therapy Reduces AIDS Progression/ Death in Individuals with Acute Opportunistic Infections: A Multicenter Randomized Strategy Trial

Andrew R. Zolopa; Janet Andersen; Lauren Komarow; Ian Sanne; Alejandro Sanchez; Evelyn Hogg; Carol Suckow; William G. Powderly

Background Optimal timing of ART initiation for individuals presenting with AIDS-related OIs has not been defined. Methods and Findings A5164 was a randomized strategy trial of “early ART” - given within 14 days of starting acute OI treatment versus “deferred ART” - given after acute OI treatment is completed. Randomization was stratified by presenting OI and entry CD4 count. The primary week 48 endpoint was 3-level ordered categorical variable: 1. Death/AIDS progression; 2. No progression with incomplete viral suppression (ie HIV viral load (VL) ≥50 copies/ml); 3. No progression with optimal viral suppression (ie HIV VL <50 copies/ml). Secondary endpoints included: AIDS progression/death; plasma HIV RNA and CD4 responses and safety parameters including IRIS. 282 subjects were evaluable; 141 per arm. Entry OIs included Pneumocytis jirovecii pneumonia 63%, cryptococcal meningitis 12%, and bacterial infections 12%. The early and deferred arms started ART a median of 12 and 45 days after start of OI treatment, respectively. The difference in the primary endpoint did not reach statistical significance: AIDS progression/death was seen in 20 (14%) vs. 34 (24%); whereas no progression but with incomplete viral suppression was seen in 54 (38%) vs. 44 (31%); and no progression with optimal viral suppression in 67 (48%) vs 63 (45%) in the early vs. deferred arm, respectively (p = 0.22). However, the early ART arm had fewer AIDS progression/deaths (OR = 0.51; 95% CI = 0.27–0.94) and a longer time to AIDS progression/death (stratified HR = 0.53; 95% CI = 0.30–0.92). The early ART had shorter time to achieving a CD4 count above 50 cells/mL (p<0.001) and no increase in adverse events. Conclusions Early ART resulted in less AIDS progression/death with no increase in adverse events or loss of virologic response compared to deferred ART. These results support the early initiation of ART in patients presenting with acute AIDS-related OIs, absent major contraindications. Trial Registration ClinicalTrials.gov NCT00055120


The Lancet | 2012

Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir versus co-formulated efavirenz, emtricitabine, and tenofovir for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3 trial, analysis of results after 48 weeks

Paul E. Sax; Edwin DeJesus; Anthony Mills; Andrew R. Zolopa; Calvin Cohen; David A. Wohl; Joel E. Gallant; Hui C. Liu; Lijie Zhong; Kitty Yale; Kirsten White; Brian P. Kearney; Javier Szwarcberg; Erin Quirk; Andrew K. Cheng

BACKGROUND The integrase inhibitor elvitegravir (EVG) has been co-formulated with the CYP3A4 inhibitor cobicistat (COBI), emtricitabine (FTC), and tenofovir disoproxil fumarate (TDF) in a single tablet given once daily. We compared the efficacy and safety of EVG/COBI/FTC/TDF with standard of care-co-formulated efavirenz (EFV)/FTC/TDF-as initial treatment for HIV infection. METHODS In this phase 3 trial, treatment-naive patients from outpatient clinics in North America were randomly assigned by computer-generated allocation sequence with a block size of four in a 1:1 ratio to receive EVG/COBI/FTC/TDF or EFV/FTC/TDF, once daily, plus matching placebo. Patients and study staff involved in giving study treatment, assessing outcomes, and collecting and analysing data were masked to treatment allocation. Eligibility criteria included screening HIV RNA concentration of 5000 copies per mL or more, and susceptibility to efavirenz, emtricitabine, and tenofovir. The primary endpoint was HIV RNA concentration of fewer than 50 copies per mL at week 48. The study is registered with ClinicalTrials.gov, number NCT01095796. FINDINGS 700 patients were randomly assigned and treated (348 with EVG/COBI/FTC/TDF, 352 with EFV/FTC/TDF). EVG/COBI/FTC/TDF was non-inferior to EFV/FTC/TDF; 305/348 (87·6%) versus 296/352 (84·1%) of patients had HIV RNA concentrations of fewer than 50 copies per mL at week 48 (difference 3·6%, 95% CI -1·6% to 8·8%). Proportions of patients discontinuing drugs for adverse events did not differ substantially (13/348 in the EVG/COBI/FTC/TDF group vs 18/352 in the EFV/FTC/TDF group). Nausea was more common with EVG/COBI/FTC/TDF than with EFV/FTC/TDF (72/348 vs 48/352) and dizziness (23/348 vs 86/352), abnormal dreams (53/348 vs 95/352), insomnia (30/348 vs 49/352), and rash (22/348 vs 43/352) were less common. Serum creatinine concentration increased more by week 48 in the EVG/COBI/FTC/TDF group than in the EFV/FTC/TDF group (median 13 μmol/L, IQR 5 to 20 vs 1 μmol/L, -6 to 8; p<0·001). INTERPRETATION If regulatory approval is given, EVG/COBI/FTC/TDF would be the only single-tablet, once-daily, integrase-inhibitor-based regimen for initial treatment of HIV infection. FUNDING Gilead Sciences.


AIDS | 2003

High levels of adherence do not prevent accumulation of HIV drug resistance mutations.

David R. Bangsberg; Edwin D. Charlebois; Robert M. Grant; Mark Holodniy; Steven G. Deeks; Sharon Perry; Kathleen Conroy; Richard A.F. Clark; David Guzman; Andrew R. Zolopa; Andrew R. Moss

Objectives: To assess the relationship between development of antiretroviral drug resistance and adherence by measured treatment duration, virologic suppression, and the rate of accumulating new drug resistance mutations at different levels of adherence. Methods: Adherence was measured with unannounced pill counts performed at the participants usual place of residence in a prospective cohort of HIV-positive urban poor individuals. Two genotypic resistance tests separated by 6 months (G1 and G2) were obtained in individuals on a stable regimen and with detectable viremia (> 50 copies/ml). The primary resistance outcome was the number of new HIV antiretroviral drug resistance mutations occurring over the 6 months between G1 and G2. Results: High levels of adherence were closely associated with greater time on treatment (P < 0.0001) and viral suppression (P < 0.0001) in 148 individuals. In a subset of 57 patients with a plasma viral load > 50 copies/ml on stable therapy, the accumulation of new drug resistance mutations was positively associated with the duration of prior treatment (P = 0.03) and pill count adherence (P = 0.002). Assuming fully suppressed individuals (< 50 copies/ml) do not develop resistance, it was estimated that 23% of all drug resistance occurs in the top quintile of adherence (92–100%), and over 50% of all drug resistance mutations occur in the top two quintiles of adherence (79–100%). Conclusion: Increasing rates of viral suppression at high levels of adherence is balanced by increasing rates of drug resistance among viremic patients. Exceptionally high levels of adherence will not prevent population levels of drug resistance.


Annals of Internal Medicine | 1999

HIV-1 genotypic resistance patterns predict response to saquinavir-ritonavir therapy in patients in whom previous protease inhibitor therapy had failed.

Andrew R. Zolopa; Robert W. Shafer; Ann Warford; Jose G. Montoya; Phillip Hsu; David Katzenstein; Thomas C. Merigan; Brad Efron

Combination antiretroviral therapy for HIV-1 infection has resulted in profound control of HIV replication in vivo, improved immune function, and significant decreases in AIDS-related morbidity and mortality (1-9). For many persons, however, this therapy does not provide sustained viral suppression or durable clinical benefit (10, 11). Potential reasons for the loss of viral suppression include host immune defects, poor adherence to therapy, pharmacologic factors, and drug resistance (10-17). However, HIV-1 resistance to drug therapy is probably the central factor in the loss of viral suppression (18-22). Mutations that result in reduced drug susceptibility have been demonstrated in vitro for all currently available antiretroviral agents, and some of these mutations have been associated with increasing plasma HIV-1 RNA levels and disease progression in clinical trials (19-30). Genotypic and phenotypic methods of measuring drug resistance are increasingly available to clinicians (31-37). However, the role of these tests in clinical practice has not been fully assessed. Many experts have been skeptical of resistance testing, although a recent consensus statement provides cautious support for testing in certain clinical circumstances (38-42). Our objective was to determine the genotypic predictors of virologic response to saquinavirritonavir combination therapy in patients in whom therapy with at least one protease inhibitor-containing antiretroviral regimen had failed. We investigated whether HIV-1 reverse transcriptase and protease genotype predicts virologic response to saquinavirritonavir by week 12 and week 26 and compared those data with predictors from clinical and antiretroviral treatment history. Methods Patients Two of the investigators treated patients in a university-based clinic that provides primary care for 500 HIV-infected adults. We identified 54 patients who received saquinavirritonavir between October 1996 and February 1998 after therapy with at least one protease inhibitor-containing antiretroviral regimen had failed. Treatment failure was defined as a greater than 0.5 log10 copies/mL (more than threefold) increase in plasma HIV RNA level from a nadir value, an HIV RNA level greater than 10 000 copies/mL, or detectable HIV RNA after the level had been below the threshold of detection (<500 copies/mL) during a therapeutic regimen for more than 12 weeks. Study patients received 400 to 600 mg of saquinavir in a hard-gel formulation (Invirase, Roche Laboratories, Nutley, New Jersey) and 300 to 400 mg of ritonavir in capsule form (Norvir, Abbott Laboratories, Abbott Park, Illinois) twice daily. In addition to the two protease inhibitors, 47 patients (87%) received two nucleoside reverse transcriptase inhibitors, 4 received three nucleoside reverse transcriptase inhibitors, 2 received two nucleoside reverse transcriptase inhibitors and either nevirapine or delavirdine, and one patient received lamivudine. Clinical and demographic variables were abstracted from the medical records. Adherence, as recorded in the patients record, was categorized by the self-reported number of missed doses in the month before evaluation and was classified as none, one to two, three to seven, or eight or more. Plasma levels of HIV-1 RNA were monitored on average every 4 to 6 weeks, and samples were stored at 70 C. The Stanford University Panel on Medical Human Subjects approved this study (#M1272). HIV Genotyping Baseline HIV-1 genotype was evaluated in plasma specimens that were stored within 1 month before initiation of saquinavirritonavir therapy and were obtained while patients were still receiving an ineffective antiretroviral regimen. Plasma HIV-1 RNA was extracted, and nested polymerase chain reaction (PCR) amplification generated a 1.3-kb fragment encompassing protease and the first 750 nucleotides of reverse transcriptase (43, 44). Direct bidirectional dideoxynucleotide terminator cycle sequencing of the PCR product was performed as described elsewhere (44). Sequencing reactions were analyzed by using an ABI 377 instrument (Perkin-Elmer, Foster City, California) and were manually proofread and edited. Sequences were compared to the HIV-1 clade B consensus sequence (Los Alamos database), and differences in amino acid sequence, including positions that contained a mixture of wild-type and mutant residues, were classified as mutations (45). Phylogenetic analysis of HIV-1 RNA sequence verified lack of cross-contamination (data not shown). A priori, we decided to assess reverse transcriptase codons 41, 67, 69, 70, 74, 75, 151, 184, 210, 215, and 219 as predictors of virologic response. Mutations at these codons are known to be associated with resistance to one of the nucleoside reverse transcriptase inhibitors (25, 45). In the protease gene, mutations at codons 30, 46, 48, 54, 82, 84, and 90 were evaluated as potential predictors. We chose these major mutations a priori because they are associated with in vitro resistance to a protease inhibitor or occur commonly in patients in whom therapy with currently licensed protease inhibitors is failing. We also evaluated all other protease codons as predictors of response. Virologic Outcomes Virologic response to saquinavirritonavir was measured at two time points between 3 and 18 weeks and again around week 24 (range, 22 to 36 weeks); the median time points of the three follow-up evaluations were 4, 12, and 26 weeks. Levels of HIV-1 RNA were measured by using the Amplicor HIV Monitor Assay (Roche Molecular Systems, Alameda, California). Specimens with HIV RNA below the level of detection (<500 copies/mL) on this assay were retested by using the ultrasensitive modification with a lower limit of detection of less than 50 copies/mL (43). Virologic response to saquinavirritonavir therapy was categorized on the basis of the larger response from baseline to the first or second evaluation [median, 4 and 12 weeks]. The ordinal categories were 1) complete response if the plasma HIV-1 RNA level was less than 500 copies/mL, 2) partial response if the reduction from baseline RNA level was 0.5 log10 copies/mL or more but was not less than 500 copies/mL, and 3) nonresponse if reduction from baseline values was less than 0.5 log10 copies/mL. Statistical Analysis Demographic, clinical, and genotypic variables were analyzed as potential predictors of virologic response by using bivariate linear regression and multivariable linear regression. In the multivariable models, we included a subset of the reverse transcriptase mutations (listed above) identified through stepwise regression as significant (P<0.05) predictors. This subset of mutations was included in the model as a signed-sum variable. For the protease mutations, we included the signed sum of the seven major mutations listed above and the signed sum of three additional mutations at codons 10, 19, and 71, which were found to be statistically significant bivariate predictors. The signed-sum variable is derived by a summation of the relevant mutations identified in the baseline sequence. A separate sum is derived from the seven major protease mutations, the three additional protease mutations, and the subset of statistically significant reverse transcriptase mutations (codons 69 and 210). In the signed-sum variable, mutations that are positively associated with virologic outcome (such as protease mutation D30N) are assigned a positive sign (+1) and mutations negatively associated with outcome are assigned a negative sign (1). We used the Cook distance to assess skewing of the ordinal outcome variable in the final multivariable model (model 5, Table 3). The value of 0.11 indicated no significant skewing; this result supports the use of linear regression models (46). We also evaluated the multivariate models for bias that would result from overfitting of the data. We used a bootstrap technique to estimate bias (optimism) in the explained variance values (R 2) for the models presented and found minimal upward bias; for example, model 3 in Table 3 has a bias of approximately one fifth of the SE (data for other models not shown) (47). A bootstrap technique was used to provide the 95% CI estimates for the R 2 values in Table 3. We selected 25 bootstrap samples of 54 with replacement from the original 54 patients to estimate the 95% CIs. The Wilcoxon rank test was used for comparisons between specific previous protease inhibitors in Table 1, and the F test was used for comparisons between models in Table 3. Two-sided P values are reported for all analyses. All analyses were conducted by using S-PLUS, version 4.0 (MathSoft, Seattle, Washington). Table 1. Baseline Demographics, Clinical Characteristics, and Antiretroviral Treatment History as Predictors of Virologic Response to saquinavirritonavir Therapy Table 2. Protease Mutation Patterns at Baseline and Response to saquinavirritonavir Combination Therapy Table 3. Multivariable Linear Regression Models of Clinical, Antiretroviral Treatment History, and HIV-1 Genotypic Predictors of Virologic Response by Week 12 of saquinavirritonavir Therapy Results Virologic Response to saquinavirritonavir Therapy Of the 54 study patients, 22 (41%) achieved a complete response, with plasma HIV-1 RNA levels less than 500 copies/mL by the second follow-up evaluation (at a median of 12 weeks). Of these 22 patients, 10 (18.5% of the entire cohort) achieved a plasma HIV-1 RNA level less than 50 copies/mL. Fourteen patients (26%) had a partial response to saquinavirritonavir, and 18 (33%) were nonresponders (Table 1). The virologic response to saquinavirritonavir is shown by initial response category in Figure 1. The response waned somewhat in the partial and complete response groups by week 26: The HIV RNA level remained below 500 copies/mL in 15 patients (28%) and below 50 copies/mL in 10 patients (19%). Figure 1. Virologic response to saquinavir plus ritonavir through week 26 based on response by week 12. Pred


The Journal of Infectious Diseases | 2007

Phase 2 Study of the Safety and Efficacy of Vicriviroc, a CCR5 Inhibitor, in HIV-1-Infected, Treatment-Experienced Patients: AIDS Clinical Trials Group 5211

Roy M. Gulick; Zhaohui Su; Charles Flexner; Michael D. Hughes; Paul R. Skolnik; Timothy Wilkin; Robert Gross; Amy Krambrink; Eoin Coakley; Wayne Greaves; Andrew R. Zolopa; Richard C. Reichman; Catherine Godfrey; Martin S. Hirsch; Daniel R. Kuritzkes

BACKGROUND Vicriviroc, an investigational CCR5 inhibitor, demonstrated short-term antiretroviral activity in a phase 1 study. METHODS The present study was a double-blind, randomized phase 2 study of vicriviroc in treatment-experienced, human immunodeficiency virus (HIV)-infected subjects experiencing virologic failure while receiving a ritonavir-containing regimen with an HIV-1 RNA level >or=5000 copies/mL and CCR5-using virus. Vicriviroc at 5, 10, or 15 mg or placebo was added to the failing regimen for 14 days, after which the antiretroviral regimen was optimized. The primary end point was the change in plasma HIV-1 RNA levels at day 14; secondary end points included safety/tolerability and HIV-1 RNA changes at week 24. RESULTS One hundred eighteen subjects were randomized with a median HIV-1 RNA level of 36,380 (4.56 log(10)) copies/mL and a median CD4 cell count of 146 cells/mm(3). At 14 days and 24 weeks, mean changes in HIV-1 RNA level (log(10) copies/mL) were greater in the vicriviroc groups (-0.87 and -1.51 [5 mg], -1.15 and -1.86 [10 mg], and -0.92 and -1.68 [15 mg]) than in the placebo group (+0.06 and -0.29) (P<.01). Grade 3/4 adverse events were similar across groups. Malignancies occurred in 6 subjects randomized to vicriviroc and in 2 to placebo. CONCLUSIONS In HIV-1-infected, treatment-experienced patients, vicriviroc demonstrated potent virologic suppression through 24 weeks. The relationship of vicriviroc to malignancy is uncertain. Further development of vicriviroc in treatment-experienced patients is warranted.


Journal of Virology | 2003

Mutation Patterns and Structural Correlates in Human Immunodeficiency Virus Type 1 Protease following Different Protease Inhibitor Treatments

Thomas D. Wu; Celia A. Schiffer; Jonathan Taylor; Rami Kantor; Sunwen Chou; Dennis Israelski; Andrew R. Zolopa; W. Jeffrey Fessel; Robert W. Shafer

ABSTRACT Although many human immunodeficiency virus type 1 (HIV-1)-infected persons are treated with multiple protease inhibitors in combination or in succession, mutation patterns of protease isolates from these persons have not been characterized. We collected and analyzed 2,244 subtype B HIV-1 isolates from 1,919 persons with different protease inhibitor experiences: 1,004 isolates from untreated persons, 637 isolates from persons who received one protease inhibitor, and 603 isolates from persons receiving two or more protease inhibitors. The median number of protease mutations per isolate increased from 4 in untreated persons to 12 in persons who had received four or more protease inhibitors. Mutations at 45 of the 99 amino acid positions in the protease—including 22 not previously associated with drug resistance—were significantly associated with protease inhibitor treatment. Mutations at 17 of the remaining 99 positions were polymorphic but not associated with drug treatment. Pairs and clusters of correlated (covarying) mutations were significantly more likely to occur in treated than in untreated persons: 115 versus 23 pairs and 30 versus 2 clusters, respectively. Of the 115 statistically significant pairs of covarying residues in the treated isolates, 59 were within 8 Å of each other—many more than would be expected by chance. In summary, nearly one-half of HIV-1 protease positions are under selective drug pressure, including many residues not previously associated with drug resistance. Structural factors appear to be responsible for the high frequency of covariation among many of the protease residues. The presence of mutational clusters provides insight into the complex mutational patterns required for HIV-1 protease inhibitor resistance.


Journal of Acquired Immune Deficiency Syndromes | 2010

Inflammation and Mortality in HIV-Infected Adults: Analysis of the FRAM Study Cohort

Phyllis C. Tien; Andy I. Choi; Andrew R. Zolopa; Constance A. Benson; Russell P. Tracy; Rebecca Scherzer; Peter Bacchetti; Michael G. Shlipak; Carl Grunfeld

Objective: To determine the association of inflammatory markers, fibrinogen, and C-reactive protein (CRP), with 5-year mortality risk. Methods: Vital status was ascertained in 922 HIV-infected participants from the Study of Fat Redistribution and Metabolic Change in HIV infection. Multivariable logistic regression estimated odds ratios after adjustment for demographic, cardiovascular, and HIV-related factors. Results: Over a 5-year period, HIV-infected participants with fibrinogen levels in the highest tertile (>406 mg/dL) had 2.6-fold higher adjusted odds of death than those with fibrinogen in the lowest tertile (<319 mg/dL). Those with high CRP (>3 mg/L) had 2.7-fold higher adjusted odds of death than those with CRP <1 mg/L. When stratified by CD4 count category, fibrinogen (as a linear variable) remained independently associated [odds ratio (95% confidence intervals)] per 100 mg/dL increase in fibrinogen: 1.93 (1.57 to 2.37); 1.43 (1.14 to 1.79); 1.43 (1.14 to 1.81); and 1.30 (1.04 to 1.63) for CD4 <200, 200-350, >350 to 500, and >500 cells per microliter, respectively. Higher CRP also remained associated with higher odds of death overall and within each CD4 subgroup. Conclusions: Fibrinogen and CRP are strong and independent predictors of mortality in HIV-infected adults. Our findings suggest that even in those with relatively preserved CD4 counts >500 cells per microliter, inflammation remains an important risk factor for mortality. Further investigation should determine whether interventions to reduce inflammation might decrease mortality risk in HIV-infected individuals.


Journal of Acquired Immune Deficiency Syndromes | 2014

Tenofovir alafenamide vs. tenofovir disoproxil fumarate in single tablet regimens for initial HIV-1 therapy: A randomized phase 2 study

Paul E. Sax; Andrew R. Zolopa; Indira Brar; Richard Elion; Roberto Ortiz; Frank Post; Hui Wang; Christian Callebaut; Hal Martin; Marshall Fordyce; Scott McCallister

Objectives:To evaluate the safety and efficacy of the novel tenofovir prodrug, tenofovir alafenamide (TAF), as part of a single-tablet regimen (STR) for the initial treatment of HIV-1 infection. Design:Phase 2, randomized, double-blind, double-dummy, multicenter, active-controlled study. Methods:Antiretroviral naive adults with HIV-1 RNA ≥5000 copies per milliliter and a CD4 count ≥50 cells per microliter were randomized 2:1 to receive an STR of elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide (E/C/F/TAF) or elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate (E/C/F/TDF), plus placebo for 48 weeks. Results:Patients on both E/C/F/TAF (n = 112) and E/C/F/TDF (n = 58) had high rates of virologic suppression (<50 HIV copies per milliliter) at week 24 (86.6%; 89.7%) and at week 48 (88.4%; 87.9%), and had similar improvements in CD4 at week 48 (177; 204), respectively. Both treatments were well tolerated, and most adverse events were self-limiting and of mild to moderate severity. Compared with patients on E/C/F/TDF, patients on E/C/F/TAF had smaller reductions in estimated creatinine clearance (−5.5 vs. −10.1 mL/min, P = 0.041), significantly less renal tubular proteinuria, and smaller changes in bone mineral density for hip (−0.62% vs. −2.39%, P < 0.001) and spine (−1.00% vs. −3.37%, P < 0.001). Patients on E/C/F/TAF had higher increases in total cholesterol, low-density lipoprotein, and high-density lipoprotein, but the total cholesterol/high-density lipoprotein ratio was unchanged for both. Conclusions:Treatment-naive patients given the STR that contained either TAF or TDF achieved a high rate of virologic success. Compared with those receiving TDF, patients on E/C/F/TAF experienced significantly smaller changes in estimated creatinine clearance, renal tubular proteinuria, and bone mineral density.

Collaboration


Dive into the Andrew R. Zolopa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul E. Sax

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew R. Moss

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel E. Gallant

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge